Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 90(12): 5549-5560, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27009955

RESUMO

UNLABELLED: Inflammation may be maladaptive to the control of viral infection when it impairs interferon (IFN) responses, enhancing viral replication and spread. Dysregulated immunity as a result of inappropriate innate inflammatory responses is a hallmark of chronic viral infections such as, hepatitis B virus and hepatitis C virus (HCV). Previous studies from our laboratory have shown that expression of an IFN-stimulated gene (ISG), ubiquitin-like protease (USP)18 is upregulated in chronic HCV infection, leading to impaired hepatocyte responses to IFN-α. We examined the ability of inflammatory stimuli, including tumor necrosis factor alpha (TNF-α), lipopolysaccharide (LPS), interleukin-6 (IL-6) and IL-10 to upregulate hepatocyte USP18 expression and blunt the IFN-α response. Human hepatoma cells and primary murine hepatocytes were treated with TNF-α/LPS/IL-6/IL-10 and USP18, phosphorylated (p)-STAT1 and myxovirus (influenza virus) resistance 1 (Mx1) expression was determined. Treatment of Huh7.5 cells and primary murine hepatocytes with LPS and TNF-α, but not IL-6 or IL-10, led to upregulated USP18 expression and induced an IFN-α refractory state, which was reversed by USP18 knockdown. Liver inflammation was induced in vivo using a murine model of hepatic ischemia/reperfusion injury. Hepatic ischemia/reperfusion injury led to an induction of USP18 expression in liver tissue and promotion of lymphocytic choriomeningitis replication. These data demonstrate that certain inflammatory stimuli (TNF-α and LPS) but not others (IL-6 and IL-10) target USP18 expression and thus inhibit IFN signaling. These findings represent a new paradigm for how inflammation alters hepatic innate immune responses, with USP18 representing a potential target for intervention in various inflammatory states. IMPORTANCE: Inflammation may prevent the control of viral infection when it impairs the innate immune response, enhancing viral replication and spread. Blunted immunity as a result of inappropriate innate inflammatory responses is a common characteristic of chronic viral infections. Previous studies have shown that expression of certain interferon-stimulated genes is upregulated in chronic HCV infection, leading to impaired hepatocyte responses. In this study, we show that multiple inflammatory stimuli can modulate interferon stimulated gene expression and thus inhibit hepatocyte interferon signaling via USP18 induction. These findings represent a new paradigm for how inflammation alters hepatic innate immune responses, with the induction of USP18 representing a potential target for intervention in various inflammatory states.


Assuntos
Endopeptidases/genética , Hepatócitos/imunologia , Hepatócitos/metabolismo , Interferon-alfa/metabolismo , Lipopolissacarídeos/farmacologia , Fígado/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitina Tiolesterase/genética , Animais , Linhagem Celular Tumoral , Endopeptidases/metabolismo , Hepacivirus/fisiologia , Hepatite C Crônica/imunologia , Hepatócitos/virologia , Humanos , Imunidade Inata , Inflamação/virologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interleucina-10/farmacologia , Interleucina-6/farmacologia , Isquemia/sangue , Fígado/irrigação sanguínea , Fígado/lesões , Fígado/patologia , Camundongos , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Ubiquitina Tiolesterase/metabolismo
2.
Nat Mater ; 15(11): 1212-1221, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27525571

RESUMO

The liver and spleen are major biological barriers to translating nanomedicines because they sequester the majority of administered nanomaterials and prevent delivery to diseased tissue. Here we examined the blood clearance mechanism of administered hard nanomaterials in relation to blood flow dynamics, organ microarchitecture and cellular phenotype. We found that nanomaterial velocity reduces 1,000-fold as they enter and traverse the liver, leading to 7.5 times more nanomaterial interaction with hepatic cells relative to peripheral cells. In the liver, Kupffer cells (84.8 ± 6.4%), hepatic B cells (81.5 ± 9.3%) and liver sinusoidal endothelial cells (64.6 ± 13.7%) interacted with administered PEGylated quantum dots, but splenic macrophages took up less material (25.4 ± 10.1%) due to differences in phenotype. The uptake patterns were similar for two other nanomaterial types and five different surface chemistries. Potential new strategies to overcome off-target nanomaterial accumulation may involve manipulating intra-organ flow dynamics and modulating the cellular phenotype to alter hepatic cell interactions.


Assuntos
Fígado/metabolismo , Nanoestruturas , Dureza , Fígado/citologia , Fenótipo , Propriedades de Superfície
3.
J Virol ; 88(11): 6195-204, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24648452

RESUMO

UNLABELLED: Coronaviruses express a deubiquitinating protein, the papain-like protease-2 (PLP2), that removes both ubiquitin and the ubiquitin-like interferon (IFN)-stimulated gene 15 (ISG15) protein from target proteins. ISG15 has antiviral activity against a number of viruses; therefore, we examined the effect of ISG15 conjugation (ISGylation) in a model of acute viral hepatitis induced by the murine hepatitis virus strain 3 (MHV-3) coronavirus. Mice deficient in the ISG15 deconjugating enzyme, ubiquitin-specific peptidase-18 (USP18), accumulate high levels of ISG15-conjugated proteins and are hypersensitive to type I IFN. Infecting USP18(-/-) mice with MHV-3 resulted in extended survival (8 ± 1.2 versus 4 days) and in improved liver histology, a decreased inflammatory response, and viral titers 1 to 2 logs lower than in USP18(+/+) mice. The suppression of viral replication was not due to increased IFN since infected USP18(-/-) mice had neither increased hepatic IFN-α, -ß, or -γ mRNA nor circulating protein. Instead, delayed MHV-3 replication coincided with high levels of cellular ISGylation. Decreasing ISGylation by knockdown of the ISG15 E1 enzyme, Ube1L, in primary USP18(+/+) and USP18(-/-) hepatocytes led to increased MHV-3 replication. Both in vitro and in vivo, increasing MHV-3 titers were coincident with increased PLP2 mRNA and decreased ISGylation over the course of infection. The pharmacologic inhibition of the PLP2 enzyme in vitro led to decreased MHV-3 replication. Overall, these results demonstrate the antiviral effect of ISGylation in an in vivo model of coronavirus-induced mouse hepatitis and illustrate that PLP2 manipulates the host innate immune response through the ISG15/USP18 pathway. IMPORTANCE: There have been a number of serious worldwide pandemics due to widespread infections by coronavirus. This virus (in its many forms) is difficult to treat, in part because it is very good at finding "holes" in the way that the host (the infected individual) tries to control and eliminate the virus. In this study, we demonstrate that an important host viral defense-the ISG15 pathway-is only partially effective in controlling severe coronavirus infection. Activation of the pathway is very good at suppressing viral production, but over time the virus overwhelms the host response and the effects of the ISG15 pathway. These data provide insight into host-virus interactions during coronavirus infection and suggest that the ISG15 pathway is a reasonable target for controlling severe coronavirus infection although the best treatment will likely involve multiple pathways and targets.


Assuntos
Infecções por Coronavirus/metabolismo , Citocinas/metabolismo , Hepatite Viral Animal/metabolismo , Vírus da Hepatite Murina , Papaína/metabolismo , Ubiquitina Tiolesterase/deficiência , Alanina Transaminase/sangue , Análise de Variância , Animais , Aspartato Aminotransferases/sangue , Western Blotting , Proteases Semelhantes à Papaína de Coronavírus , Primers do DNA/genética , Hepatite Viral Animal/virologia , Hepatócitos , Interferons/sangue , Macrófagos Peritoneais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Papaína/antagonistas & inibidores , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sobrevida , Enzimas Ativadoras de Ubiquitina/metabolismo , Ubiquitinas/metabolismo
4.
Genomics ; 102(5-6): 491-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24100145

RESUMO

We identified 7 SHP-1 (PTPN6) transcripts using epithelial cancer-derived cell lines. Four were shown to utilize the epithelial promoter 1 to transcribe a full-length, a partial (exon 3) or complete (exons 3 & 4) deletion of the N-SH2 domain, and also a non-coding transcript having a stop codon caused by a frame shift due to intron 2 retention. Three additional transcripts were shown to utilize the hematopoietic promoter 2 to transcribe a full-length, a partial (exon 3) deletion of the N-SH2 domain and a non-coding transcript with intron 2 retention. We show that endogenous proteins corresponding to the open-reading-frame (ORF) transcripts are produced. Using GST-fusion proteins we show that each product of the ORF SHP-1 transcripts has phosphatase activity and isoforms with an N-SH2 deletion have increased phosphatase activity and novel protein-protein interactions. This study is the first to document utilization of promoter 2 by SHP-1 transcripts and a noncoding transcript in human epithelial cells.


Assuntos
Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Processamento Alternativo , Linhagem Celular Tumoral , Éxons , Mutação da Fase de Leitura , Células HEK293 , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Células Jurkat , Células MCF-7 , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 6/química , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
5.
iScience ; 26(11): 108213, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38026201

RESUMO

The large size and vascular accessibility of the laboratory rat (Rattus norvegicus) make it an ideal hepatic animal model for diseases that require surgical manipulation. Often, the disease susceptibility and outcomes of inflammatory pathologies vary significantly between strains. This study uses single-cell transcriptomics to better understand the complex cellular network of the rat liver, as well as to unravel the cellular and molecular sources of inter-strain hepatic variation. We generated single-cell and single-nucleus transcriptomic maps of the livers of healthy Dark Agouti and Lewis rat strains and developed a factor analysis-based bioinformatics analysis pipeline to study data covariates, such as strain and batch. Using this approach, we discovered transcriptomic variation within the hepatocyte and myeloid populations that underlie distinct cell states between rat strains. This finding will help provide a reference for future investigations on strain-dependent outcomes of surgical experiment models.

6.
Lab Invest ; 92(9): 1285-96, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22732935

RESUMO

The outbreak of severe acute respiratory syndrome (SARS) in 2003 reinforces the potential of lethal pandemics of respiratory viral infections. The underlying mechanisms of SARS are still largely undefined. Long pentraxin PTX3, a humoral mediator of innate immunity, has been reported to have anti-viral effects. We examined the role of PTX3 in coronavirus murine hepatitis virus strain 1 (MHV-1)-induced acute lung injury, a previously reported animal model for SARS. PTX3-deficient mice (129/SvEv/C57BL6/J) and their wild-type (WT) littermates were intranasally infected MHV-1. These mice were also treated with recombinant PTX3. Effects of PTX3 on viral binding and infectivity were determined in vitro. Cytokine expression, severity of lung injury, leukocyte infiltration and inflammatory responses were examined in vivo. In PTX3 WT mice, MHV-1 induced PTX3 expression in the lung and serum in a time-dependent manner. MHV-1 infection led to acute lung injury with greater severity in PTX3-deficient mice than that in WT mice. PTX3 deficiency enhanced early infiltration of neutrophils and macrophages in the lung. PTX3 bound to MHV-1 and MHV-3 and reduced MHV-1 infectivity in vitro. Administration of recombinant PTX3 significantly accelerated viral clearance in the lung, attenuated MHV-1-induced lung injury, and reduced early neutrophil influx and elevation of inflammatory mediators in the lung. Results from this study indicate a protective role of PTX3 in coronaviral infection-induced acute lung injury.


Assuntos
Proteína C-Reativa/fisiologia , Modelos Animais de Doenças , Proteínas do Tecido Nervoso/fisiologia , Síndrome Respiratória Aguda Grave/prevenção & controle , Animais , Proteína C-Reativa/genética , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Síndrome Respiratória Aguda Grave/virologia , Carga Viral
7.
Transfusion ; 52(8): 1799-805, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22257295

RESUMO

BACKGROUND: Sialylation of the N-linked glycan on asparagine-297 within the Fc region of intravenous gammaglobulins (IVIgs) was shown to be necessary for efficacy of IVIg in the amelioration of experimental inflammatory arthritis. To test the role for Fc sialylation of IVIg in immune modulating therapies beyond the K/BxN arthritis model, we examined the efficacy of sialylated compared to nonsialylated IVIg for the ability to attenuate immune thrombocytopenia (ITP) in a mouse model that approximates the clinical setting of human ITP. STUDY DESIGN AND METHODS: We used a published, passive anti-platelet (PLT) dose-escalation mouse model of ITP that approximates clinical ITP. PLT counts were followed over time before and after IVIg treatment. IVIg from two different manufacturers was used to prepare untreated and neuraminidase-treated IVIg. Sambucus nigra agglutinin (SNA) affinity chromatography was used to obtain sialic acid-enriched and -depleted IVIg. Sialic acid content was determined using Western blot, enzyme-linked immunosorbent assay, and high-performance liquid chromatography. RESULTS: Results were the same using sialylated and desialylated (neuraminidase-treated) IVIg from two different manufacturers. No differences were observed between sialic acid-enriched and -depleted IVIg compared to normal IVIg in their efficacy to alleviate ITP. Using quantitative reverse transcription-polymerase chain reaction, no increase in the spleen FcγRIIB mRNA was detectable, but a pronounced increase of FcγRIIB mRNA in the marrow was seen after IVIg administration. CONCLUSIONS: We conclude that IVIg ameliorates experimental ITP by a mechanism that is independent of sialylation either in the Fc or the Fab region of IVIg.


Assuntos
Imunoglobulinas Intravenosas/farmacologia , Fatores Imunológicos/farmacologia , Ácido N-Acetilneuramínico/metabolismo , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Púrpura Trombocitopênica Idiopática/imunologia , Animais , Asparagina/metabolismo , Modelos Animais de Doenças , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Lectinas de Plantas/farmacologia , Contagem de Plaquetas , Polissacarídeos/metabolismo , Púrpura Trombocitopênica Idiopática/metabolismo , RNA Mensageiro/metabolismo , Receptores de IgG/genética , Proteínas Inativadoras de Ribossomos/farmacologia
8.
Nucleic Acids Res ; 38(19): 6684-96, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20558599

RESUMO

CD200, a type I membrane glycoprotein, plays an important role in prevention of inflammatory disorders, graft rejection, autoimmune diseases and spontaneous fetal loss. It also regulates tumor immunity. A truncated CD200 (CD200(tr)) resulting from alternative splicing has been identified and characterized as a functional antagonist to full-length CD200. Thus, it is important to explore the mechanism(s) controlling alternative splicing of CD200. In this study, we identified an exonic splicing enhancer (ESE) located in exon 2, which is a putative binding site for a splicing regulatory protein SF2/ASF. Deletion or mutation of the ESE site decreased expression of the full-length CD200. Direct binding of SF2/ASF to the ESE site was confirmed by RNA electrophoretic mobility shift assay (EMSA). Knockdown of expression of SF2/ASF resulted in the same splicing pattern as seen after deletion or mutation of the ESE, whereas overexpression of SF2/ASF increased expression of the full-length CD200. In vivo studies showed that viral infection reversed the alternative splicing pattern of CD200 with increased expression of SF2/ASF and the full-length CD200. Taken together, our data suggest for the first time that SF2/ASF regulates the function of CD200 by controlling CD200 alternative splicing, through direct binding to an ESE located in exon 2 of CD200.


Assuntos
Processamento Alternativo , Antígenos CD/genética , Éxons , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Ribonucleico , Animais , Antígenos CD/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Infecções por Coronavirus/genética , Feminino , Humanos , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina , Mutação , Fatores de Processamento de Serina-Arginina
9.
Hepatol Commun ; 6(4): 821-840, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34792289

RESUMO

The critical functions of the human liver are coordinated through the interactions of hepatic parenchymal and non-parenchymal cells. Recent advances in single-cell transcriptional approaches have enabled an examination of the human liver with unprecedented resolution. However, dissociation-related cell perturbation can limit the ability to fully capture the human liver's parenchymal cell fraction, which limits the ability to comprehensively profile this organ. Here, we report the transcriptional landscape of 73,295 cells from the human liver using matched single-cell RNA sequencing (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq). The addition of snRNA-seq enabled the characterization of interzonal hepatocytes at a single-cell resolution, revealed the presence of rare subtypes of liver mesenchymal cells, and facilitated the detection of cholangiocyte progenitors that had only been observed during in vitro differentiation experiments. However, T and B lymphocytes and natural killer cells were only distinguishable using scRNA-seq, highlighting the importance of applying both technologies to obtain a complete map of tissue-resident cell types. We validated the distinct spatial distribution of the hepatocyte, cholangiocyte, and mesenchymal cell populations by an independent spatial transcriptomics data set and immunohistochemistry. Conclusion: Our study provides a systematic comparison of the transcriptomes captured by scRNA-seq and snRNA-seq and delivers a high-resolution map of the parenchymal cell populations in the healthy human liver.


Assuntos
Fígado , Análise de Célula Única , Núcleo Celular/genética , Humanos , Análise de Sequência de RNA , Transcriptoma/genética
10.
J Virol ; 84(23): 12419-28, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20861244

RESUMO

Ubiquitination is a critical regulator of the host immune response to viral infection, and many viruses, including coronaviruses, encode proteins that target the ubiquitination system. To explore the link between coronavirus infection and the ubiquitin system, we asked whether protein degradation by the 26S proteasome plays a role in severe coronavirus infections using a murine model of SARS-like pneumonitis induced by murine hepatitis virus strain 1 (MHV-1). In vitro, the pretreatment of peritoneal macrophages with inhibitors of the proteasome (pyrrolidine dithiocarbamate [PDTC], MG132, and PS-341) markedly inhibited MHV-1 replication at an early step in its replication cycle, as evidenced by inhibition of viral RNA production. Proteasome inhibition also blocked viral cytotoxicity in macrophages, as well as the induction of inflammatory mediators such as IP-10, gamma interferon (IFN-γ), and monocyte chemoattractant protein 1 (MCP-1). In vivo, intranasal inoculation of MHV-1 results in a lethal pneumonitis in A/J mice. Treatment of A/J mice with the proteasome inhibitor PDTC, MG132, or PS-341 led to 40% survival (P < 0.01), with a concomitant improvement of lung histology, reduced pulmonary viral replication, decreased pulmonary STAT phosphorylation, and reduced pulmonary inflammatory cytokine expression. These data demonstrate that inhibition of the cellular proteasome attenuates pneumonitis and cytokine gene expression in vivo by reducing MHV-1 replication and the resulting inflammatory response. The results further suggest that targeting the proteasome may be an effective new treatment for severe coronavirus infections.


Assuntos
Infecções por Coronavirus/imunologia , Regulação da Expressão Gênica/imunologia , Vírus da Hepatite Murina/imunologia , Pneumonia/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Northern Blotting , Western Blotting , Ácidos Borônicos/farmacologia , Bortezomib , Infecções por Coronavirus/metabolismo , Citocinas/metabolismo , Primers do DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas Histológicas , Leupeptinas/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Fosforilação , Pneumonia/metabolismo , Pneumonia/virologia , Prolina/análogos & derivados , Prolina/farmacologia , Inibidores de Proteassoma , Pirazinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição STAT/metabolismo , Análise de Sobrevida , Tiocarbamatos/farmacologia , Ubiquitinação
11.
J Virol ; 84(18): 9278-91, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20631137

RESUMO

We report here investigation into the genetic basis of mouse hepatitis virus strain 1 (MHV-1) pneumovirulence. Sequencing of the 3' one-third of the MHV-1 genome demonstrated that the genetic organization of MHV-1 was similar to that of other strains of MHV. The hemagglutinin esterase (HE) protein was truncated, and reverse transcription-PCR (RT-PCR) studies confirmed previous work that suggested that the MHV-1 HE is a pseudogene. Targeted recombination was used to select chimeric viruses containing either the MHV-1 S gene or genes encoding all of the MHV-1 structural proteins, on an MHV-A59 background. Challenge studies in mice demonstrated that expression of the MHV-1 S gene within the MHV-A59 background (rA59/S(MHV-1)) increased the pneumovirulence of MHV-A59, and mice infected with this recombinant virus developed pulmonary lesions that were similar to those observed with MHV-1, although rA59/S(MHV-1) was significantly less virulent. Chimeras containing all of the MHV-1 structural genes on an MHV-A59 background were able to reproduce the severe acute respiratory syndrome (SARS)-like pathology observed with MHV-1 and reproducibly increased pneumovirulence relative to rA59/S(MHV-1), but were still much less virulent than MHV-1. These data suggest that important determinants of pneumopathogenicity are contained within the 3' one-third of the MHV-1 genome, but additional important virulence factors must be encoded in the genome upstream of the S gene. The severity of the pulmonary lesions observed correlates better with elevated levels of inflammatory cytokines than with viral replication in the lungs, suggesting that pulmonary disease has an important immunological component.


Assuntos
Pulmão/patologia , Pulmão/virologia , Glicoproteínas de Membrana/fisiologia , Vírus da Hepatite Murina/patogenicidade , Proteínas do Envelope Viral/fisiologia , Fatores de Virulência/fisiologia , Animais , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Feminino , Ordem dos Genes , Genes Virais , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/genética , Pneumonia Viral/patologia , Pneumonia Viral/virologia , RNA Viral/química , RNA Viral/genética , Recombinação Genética , Análise de Sequência de DNA , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/genética , Fatores de Virulência/genética
12.
Blood ; 113(20): 4980-91, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19139081

RESUMO

Several human histo-blood groups are glycosphingolipids, including P/P1/P(k). Glycosphingolipids are implicated in HIV-host-cell-fusion and some bind to HIV-gp120 in vitro. Based on our previous studies on Fabry disease, where P(k) accumulates and reduces infection, and a soluble P(k) analog that inhibits infection, we investigated cell surface-expressed P(k) in HIV infection. HIV-1 infection of peripheral blood-derived mononuclear cells (PBMCs) from otherwise healthy persons, with blood group P(1)(k), where P(k) is overexpressed, or blood group p, that completely lacks P(k), were compared with draw date-matched controls. Fluorescence-activated cell sorter analysis and/or thin layer chromatography were used to verify P(k) levels. P(1)(k) PBMCs were highly resistant to R5 and X4 HIV-1 infection. In contrast, p PBMCs showed 10- to 1000-fold increased susceptibility to HIV-1 infection. Surface and total cell expression of P(k), but not CD4 or chemokine coreceptor expression, correlated with infection. P(k) liposome-fused cells and CD4(+) HeLa cells manipulated to express high or low P(k) levels confirmed a protective effect of P(k). We conclude that P(k) expression strongly influences susceptibility to HIV-1 infection, which implicates P(k) as a new endogenous cell-surface factor that may provide protection against HIV-1 infection.


Assuntos
Citoproteção/imunologia , Infecções por HIV/sangue , Infecções por HIV/imunologia , HIV-1 , Triexosilceramidas/fisiologia , Antígenos CD4/metabolismo , Células Cultivadas , Citoproteção/genética , Galactosiltransferases/antagonistas & inibidores , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Predisposição Genética para Doença , Infecções por HIV/genética , HIV-1/fisiologia , Células HeLa , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Células Jurkat , RNA Interferente Pequeno/farmacologia , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Transfecção , Triexosilceramidas/metabolismo
13.
Hepatology ; 49(2): 387-97, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19085958

RESUMO

UNLABELLED: Fulminant viral hepatitis (FH) remains an important clinical problem in which the underlying pathogenesis is not well understood. Here, we present insight into the immunological mechanisms involved in FH caused by murine hepatitis virus strain 3 (MHV-3), indicating a critical role for CD4(+)CD25(+) regulatory T cells (Tregs) and production of the novel Treg effector molecule FGL2. Before infection with MHV-3, susceptible BALB/cJ mice had increased numbers of Tregs and expression of fgl2 messenger RNA (mRNA) and FGL2 protein compared with resistant A/J mice. After MHV-3 infection, plasma levels of FGL2 in BALB/cJ mice were significantly increased, correlating with increased percentage of Tregs. Treatment with anti-FGL2 antibody completely inhibited Treg activity and protected susceptible BALB/cJ mice against MHV-3-liver injury and mortality. Adoptive transfer of wild-type Tregs into resistant fgl2(-/-) mice increased their mortality caused by MHV-3 infection, whereas transfer of peritoneal exudate macrophages had no adverse effect. CONCLUSION: This study demonstrates that FGL2 is an important effector cytokine of Tregs that contributes to susceptibility to MHV-3-induced FH. The results further suggest that targeting FGL2 may lead to the development of novel treatment approaches for acute viral hepatitis infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fibrinogênio/imunologia , Hepatite Viral Animal/imunologia , Linfócitos T Reguladores/imunologia , Animais , Infecções por Coronavirus/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Terapia de Imunossupressão , Camundongos , Camundongos Endogâmicos A/imunologia , Camundongos Endogâmicos BALB C , Vírus da Hepatite Murina , Reação em Cadeia da Polimerase
14.
J Vis Exp ; (165)2020 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-33226024

RESUMO

The rat orthotopic liver transplantation (OLT) model is a powerful tool to study acute and chronic rejection. However, it is not a complete representation of human liver transplantation due to the absence of arterial reconnection. Described here is a modified transplantation procedure that includes the incorporation of hepatic artery (HA) reconnection, leading to a marked improvement in transplant outcomes. With a mean anhepatic time of 12 min and 14 s, HA reconnection results in improved perfusion of the transplanted liver and an increase in long-term recipient survival from 37.5% to 88.2%. This protocol includes the use of 3D-printed cuffs and holders to connect the portal vein and infrahepatic inferior vena cava. It can be implemented for studying multiple aspects of liver transplantation, from immune response and infection to technical aspects of the procedure. By incorporating a simple and practical method for arterial reconnection using a microvascular technique, this modified rat OLT protocol closely mimics aspects of human liver transplantation and will serve as a valuable and clinically relevant research model.


Assuntos
Rejeição de Enxerto/prevenção & controle , Artéria Hepática/cirurgia , Hepatopatias/cirurgia , Transplante de Fígado/veterinária , Veia Porta/cirurgia , Complicações Pós-Operatórias/prevenção & controle , Animais , Transplante de Fígado/métodos , Masculino , Ratos , Ratos Endogâmicos Lew
15.
ACS Nano ; 14(4): 4698-4715, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32255624

RESUMO

There is a tremendous focus on the application of nanomaterials for the treatment of cancer. Nonprimate models are conventionally used to assess the biomedical utility of nanomaterials. However, these animals often lack an intact immunological background, and the tumors in these animals do not develop spontaneously. We introduce a preclinical woodchuck hepatitis virus-induced liver cancer model as a platform for nanoparticle (NP)-based in vivo experiments. Liver cancer development in these out-bred animals occurs as a result of persistent viral infection, mimicking human hepatitis B virus-induced HCC development. We highlight how this model addresses key gaps associated with other commonly used tumor models. We employed this model to (1) track organ biodistribution of gold NPs after intravenous administration, (2) examine their subcellular localization in the liver, (3) determine clearance kinetics, and (4) characterize the identity of hepatic macrophages that take up NPs using RNA-sequencing (RNA-seq). We found that the liver and spleen were the primary sites of NP accumulation. Subcellular analyses revealed accumulation of NPs in the lysosomes of CD14+ cells. Through RNA-seq, we uncovered that immunosuppressive macrophages within the woodchuck liver are the major cell type that take up injected NPs. The woodchuck-HCC model has the potential to be an invaluable tool to examine NP-based immune modifiers that promote host anti-tumor immunity.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Animais , Modelos Animais de Doenças , Humanos , Fígado , Marmota , Distribuição Tecidual
16.
Nat Commun ; 9(1): 4383, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30348985

RESUMO

The liver is the largest solid organ in the body and is critical for metabolic and immune functions. However, little is known about the cells that make up the human liver and its immune microenvironment. Here we report a map of the cellular landscape of the human liver using single-cell RNA sequencing. We provide the transcriptional profiles of 8444 parenchymal and non-parenchymal cells obtained from the fractionation of fresh hepatic tissue from five human livers. Using gene expression patterns, flow cytometry, and immunohistochemical examinations, we identify 20 discrete cell populations of hepatocytes, endothelial cells, cholangiocytes, hepatic stellate cells, B cells, conventional and non-conventional T cells, NK-like cells, and distinct intrahepatic monocyte/macrophage populations. Together, our study presents a comprehensive view of the human liver at single-cell resolution that outlines the characteristics of resident cells in the liver, and in particular provides a map of the human hepatic immune microenvironment.


Assuntos
Fígado/citologia , Fígado/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Linfócitos B/citologia , Linfócitos B/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Monócitos/citologia , Monócitos/metabolismo , Análise de Sequência de RNA
17.
Endocrinology ; 158(2): 277-292, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27967217

RESUMO

We found previously that short-term curcumin gavage stimulated mouse hepatic fibroblast growth factor 21 (Fgf21) expression. Here we conducted mechanistic exploration and investigated the potential pathophysiological relevance on this regulation. Fgf21 stimulation was observed at messenger RNA and protein levels in mice with daily curcumin gavage for 4 or 8 days and in primary hepatocytes with curcumin treatment. Using peroxisome proliferator-activated receptor α (PPARα) agonist and antagonist, along with luciferase reporter and chromatin immune-precipitation approaches, we determined that curcumin stimulates Fgf21 transcription in a mechanism involving PPARα activation. High-fat diet (HFD) feeding also increased mouse hepatic and serum Fgf21 levels, whereas dietary curcumin intervention attenuated these increases. We found that HFD feeding reduced hepatic expression levels of genes that encode FGFR1 and ßKlotho, PGC1α, and the targets of the PPARα-PGC1α axis, whereas concomitant curcumin intervention restored or partially restored their expression levels. Importantly, hepatocytes from HFD-fed mice showed a loss of response to FGF21 treatment on Erk phosphorylation and the expression of Egr1 and cFos; this response was restored in hepatocytes from HFD-fed mice with curcumin intervention. This investigation expanded our mechanistic understanding of the metabolic beneficial effects of dietary curcumin intervention involving the regulation of Fgf21 production and the attenuation of HFD-induced Fgf21 resistance.


Assuntos
Curcumina/farmacologia , Dieta Hiperlipídica/efeitos adversos , Fatores de Crescimento de Fibroblastos/metabolismo , Fígado/efeitos dos fármacos , Animais , Curcumina/uso terapêutico , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Doenças Metabólicas/etiologia , Doenças Metabólicas/prevenção & controle , Camundongos , Obesidade/etiologia , Obesidade/prevenção & controle , PPAR alfa/metabolismo
18.
ACS Nano ; 11(3): 2428-2443, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28040885

RESUMO

A significant challenge to delivering therapeutic doses of nanoparticles to targeted disease sites is the fact that most nanoparticles become trapped in the liver. Liver-resident macrophages, or Kupffer cells, are key cells in the hepatic sequestration of nanoparticles. However, the precise role that the macrophage phenotype plays in nanoparticle uptake is unknown. Here, we show that the human macrophage phenotype modulates hard nanoparticle uptake. Using gold nanoparticles, we examined uptake by human monocyte-derived macrophages that had been driven to a "regulatory" M2 phenotype or an "inflammatory" M1 phenotype and found that M2-type macrophages preferentially take up nanoparticles, with a clear hierarchy among the subtypes (M2c > M2 > M2a > M2b > M1). We also found that stimuli such as LPS/IFN-γ rather than with more "regulatory" stimuli such as TGF-ß/IL-10 reduce per cell macrophage nanoparticle uptake by an average of 40%. Primary human Kupffer cells were found to display heterogeneous expression of M1 and M2 markers, and Kupffer cells expressing higher levels of M2 markers (CD163) take up significantly more nanoparticles than Kupffer cells expressing lower levels of surface CD163. Our results demonstrate that hepatic inflammatory microenvironments should be considered when studying liver sequestration of nanoparticles, and that modifying the hepatic microenvironment might offer a tool for enhancing or decreasing this sequestration. Our findings also suggest that models examining the nanoparticle/macrophage interaction should include studies with primary tissue macrophages.


Assuntos
Ouro/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Nanopartículas Metálicas/química , Ouro/sangue , Ouro/química , Humanos , Fígado/citologia , Macrófagos/química , Monócitos/química , Monócitos/metabolismo , Fenótipo
19.
AIDS ; 20(3): 333-43, 2006 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-16439866

RESUMO

OBJECTIVE: To determine the effect of a gp120 binding, non-cytotoxic soluble analogue of the glycosphingolipid (GSL), globotriaosyl ceramide (Gb3) on HIV infection in vitro. DESIGN: HIV-1(IIIB) (X4 virus) infection in Jurkat and phytohaemagglutinin (PHA)/interleukin-2 (IL2) activated, peripheral blood mononuclear cells (PBMC), and HIV-1(Ba-L) (R5 virus) infection of PHA activated PBMC in vitro were assessed. We monitored cell surface markers, cell viability, and viral/host cell morphology to eliminate pleiotropic effects. Viral-host cell fusion was measured to further address any inhibitory mechanism. METHODS: HIV infection was monitored by p24(gag) ELISA. CD4, CCR5, CXCR4 and apoptosis were determined by fluorescent antibody cell sorting. A model fusion system comprising a cell line transfected with either CD4 and CXCR4 or CCR5, cocultured with a cell line expressing gp120 from either X4-, R5-tropic HIV-1 or HIV-2 virions, was used. PHA/IL2 activated PBMC GSL synthesis was monitored by metabolic radiolabelling. RESULTS: AdamantylGb3 blocked X4 and R5 virus infection with a 50% inhibitory concentration of approximately 150 microM. A reverse transcriptase and a protease-resistant X4 HIV-1 strain retained adamantylGb3 sensitivity. AdamantylGb3 had minimal effect on cell viability. Treated Jurkat cells showed a small increase in CCR5/CXCR4 expression and a slight, transient CD4 down-regulation, which was probably not related to the mechanism of inhibition. Electron microscopy showed normal viral and host cell morphology following adamantylGb3 treatment, and viral entry was blocked. AdamantylGb3 was able to prevent virus-host cell fusion irrespective of HIV strain or chemokine receptor preference. CONCLUSIONS: These results suggest that adamantylGb3 may provide a new basis for blocking HIV infections, irrespective of HIV envelope/chemokine co-receptor preference or resistance to other therapeutics.


Assuntos
Antivirais/uso terapêutico , Glicolipídeos/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1 , Triexosilceramidas/uso terapêutico , Adamantano/análogos & derivados , Adamantano/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Concentração Inibidora 50 , Células Jurkat , Leucócitos Mononucleares/virologia , Microscopia Eletrônica
20.
Endocrinology ; 147(4): 1950-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16373422

RESUMO

Cdx-2 and Brn-4 are recognized as transcriptional activators for the proglucagon gene. These two homeodomain (HD) proteins are able to interact with the correspondent motifs on the G1 enhancer element of proglucagon promoter, separated by only 8 bp. We have examined Brn-4 expression in proglucagon-producing cells, isolated hamster Brn-4 cDNA, and localized its activation domain. Ectopic expression of either Cdx-2 or Brn-4 in the pancreatic B cell line In111 provoked it to express proglucagon mRNA, whereas ectopically expressing both of them further stimulated proglucagon mRNA expression in this cell line. Furthermore, Brn-4 was found to synergize with Cdx-2 in activating proglucagon promoter, and the Brn-4 activation domain was not required for this synergistic activation. When the binding site for either Cdx-2 or Brn-4 was mutated, the synergistic activation by these two HD proteins was significantly attenuated, but not abolished. We propose that both cooperative DNA binding and mutual recruitment between Cdx-2 and Brn-4 are involved in this synergistic activation and have detected physical interaction between Cdx-2 and Brn-4 by glutathione-S-transferase-fusion protein pull-down assay. Our observations suggest that Cdx-2 and Brn-4, two HD proteins that belong to two different families, exert a synergistic and redundant effect on proglucagon gene expression.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fatores do Domínio POU/fisiologia , Proglucagon/genética , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Animais , Fator de Transcrição CDX2 , Células Cultivadas , Cricetinae , DNA Complementar/isolamento & purificação , Camundongos , Proteínas do Tecido Nervoso/química , Fatores do Domínio POU/química , RNA Mensageiro/análise , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA